Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Janet M. McNicholl is active.

Publication


Featured researches published by Janet M. McNicholl.


Science | 2001

Control of a mucosal challenge and prevention of AIDS by a multiprotein DNA/MVA vaccine

Rama Rao Amara; Francois Villinger; John D. Altman; Shari L. Lydy; Shawn P. O'Neil; Silvija I. Staprans; David C. Montefiori; Yan Xu; James G. Herndon; Linda S. Wyatt; Maria Angelito Candido; Natalia Kozyr; Patricia L. Earl; James M. Smith; Hak-Ling Ma; Bennett D. Grimm; Michael L. Hulsey; Joseph D. Miller; Harold M. McClure; Janet M. McNicholl; Bernard Moss; Harriet L. Robinson

Heterologous prime/boost regimens have the potential for raising high levels of immune responses. Here we report that DNA priming followed by a recombinant modified vaccinia Ankara (rMVA) booster controlled a highly pathogenic immunodeficiency virus challenge in a rhesus macaque model. Both the DNA and rMVA components of the vaccine expressed multiple immunodeficiency virus proteins. Two DNA inoculations at 0 and 8 weeks and a single rMVA booster at 24 weeks effectively controlled an intrarectal challenge administered 7 months after the booster. These findings provide hope that a relatively simple multiprotein DNA/MVA vaccine can help to control the acquired immune deficiency syndrome epidemic.


The Lancet | 2013

Antiretroviral prophylaxis for HIV infection in injecting drug users in Bangkok, Thailand (the Bangkok Tenofovir Study): a randomised, double-blind, placebo-controlled phase 3 trial

Kachit Choopanya; Michael Martin; Pravan Suntharasamai; Udomsak Sangkum; Philip A. Mock; Manoj Leethochawalit; Sithisat Chiamwongpaet; Praphan Kitisin; Pitinan Natrujirote; Somyot Kittimunkong; Rutt Chuachoowong; Roman Gvetadze; Janet M. McNicholl; Lynn A. Paxton; Marcel E. Curlin; Craig W. Hendrix; Suphak Vanichseni

BACKGROUND Antiretroviral pre-exposure prophylaxis reduces sexual transmission of HIV. We assessed whether daily oral use of tenofovir disoproxil fumarate (tenofovir), an antiretroviral, can reduce HIV transmission in injecting drug users. METHODS In this randomised, double-blind, placebo-controlled trial, we enrolled volunteers from 17 drug-treatment clinics in Bangkok, Thailand. Participants were eligible if they were aged 20-60 years, were HIV-negative, and reported injecting drugs during the previous year. We randomly assigned participants (1:1; blocks of four) to either tenofovir or placebo using a computer-generated randomisation sequence. Participants chose either daily directly observed treatment or monthly visits and could switch at monthly visits. Participants received monthly HIV testing and individualised risk-reduction and adherence counselling, blood safety assessments every 3 months, and were offered condoms and methadone treatment. The primary efficacy endpoint was HIV infection, analysed by modified intention-to-treat analysis. This trial is registered with ClinicalTrials.gov, number NCT00119106. FINDINGS Between June 9, 2005, and July 22, 2010, we enrolled 2413 participants, assigning 1204 to tenofovir and 1209 to placebo. Two participants had HIV at enrolment and 50 became infected during follow-up: 17 in the tenofovir group (an incidence of 0·35 per 100 person-years) and 33 in the placebo group (0·68 per 100 person-years), indicating a 48·9% reduction in HIV incidence (95% CI 9·6-72·2; p=0·01). The occurrence of serious adverse events was much the same between the two groups (p=0·35). Nausea was more common in participants in the tenofovir group than in the placebo group (p=0·002). INTERPRETATION In this study, daily oral tenofovir reduced the risk of HIV infection in people who inject drugs. Pre-exposure prophylaxis with tenofovir can now be considered for use as part of an HIV prevention package for people who inject drugs. FUNDING US Centers for Disease Control and Prevention and the Bangkok Metropolitan Administration.


Journal of Virology | 2002

Intersubtype Human Immunodeficiency Virus Type 1 Superinfection following Seroconversion to Primary Infection in Two Injection Drug Users

Artur Ramos; Dale J. Hu; Lily Nguyen; Kim Oanh Phan; Suphak Vanichseni; Nattawan Promadej; Kachit Choopanya; Margaret E. Callahan; Nancy L. Young; Janet M. McNicholl; Timothy D. Mastro; Thomas M. Folks; Shambavi Subbarao

ABSTRACT In this study, we describe two cases of human immunodeficiency virus type 1 (HIV-1) intersubtype superinfection with CRF01_AE and subtype B strains, which occurred in two injection drug users participating in a prospective cohort study in Bangkok, Thailand. In both cases, the superinfecting strain was detected by molecular and serologic analyses several weeks after complete seroconversion to the primary infection with a strain belonging to a different subtype. Superinfection occurred despite specific T-cell and humoral antibody responses to the primary virus. In both cases, cross-subtype immune responses were limited or absent prior to the second infection. These data show that, in some individuals, the quality and quantity of the immune response elicited by primary HIV-1 infection may not protect against superinfection. This finding has important implications for vaccine design. HIV-1 vaccines, at a minimum, will need to include potent, broadly protective, conserved immunogens derived from several group M subtypes.


Journal of Virology | 2002

Critical Role for Env as well as Gag-Pol in Control of a Simian-Human Immunodeficiency Virus 89.6P Challenge by a DNA Prime/Recombinant Modified Vaccinia Virus Ankara Vaccine

Rama Rao Amara; James M. Smith; Silvija I. Staprans; David C. Montefiori; Francois Villinger; John D. Altman; Shawn P. O'Neil; Natalia Kozyr; Yan Xu; Linda S. Wyatt; Patricia L. Earl; James G. Herndon; Janet M. McNicholl; Harold M. McClure; Bernard Moss; Harriet L. Robinson

ABSTRACT Cellular immune responses against epitopes in conserved Gag and Pol sequences of human immunodeficiency virus type 1 have become popular targets for candidate AIDS vaccines. Recently, we used a simian-human immunodeficiency virus model (SHIV 89.6P) with macaques to demonstrate the control of a pathogenic mucosal challenge by priming with Gag-Pol-Env-expressing DNA and boosting with Gag-Pol-Env-expressing recombinant modified vaccinia virus Ankara (rMVA). Here we tested Gag-Pol DNA priming and Gag-Pol rMVA boosting to evaluate the contribution of anti-Env immune responses to viral control. The Gag-Pol vaccine raised frequencies of Gag-specific T cells similar to those raised by the Gag-Pol-Env vaccine. Following challenge, these rapidly expanded to counter the challenge infection. Despite this, the control of the SHIV 89.6P challenge was delayed and inconsistent in the Gag-Pol-vaccinated group and all of the animals underwent severe and, in most cases, sustained loss of CD4+ cells. Interestingly, most of the CD4+ cells that were lost in the Gag-Pol-vaccinated group were uninfected cells. We suggest that the rapid appearance of binding antibody for Env in Gag-Pol-Env-vaccinated animals helped protect uninfected CD4+ cells from Env-induced apoptosis. Our results highlight the importance of immune responses to Env, as well as to Gag-Pol, in the control of immunodeficiency virus challenges and the protection of CD4+ cells.


Journal of Acquired Immune Deficiency Syndromes | 2010

Trends in HIV Prevalence, Estimated HIV Incidence, and Risk Behavior Among Men Who Have Sex With Men in Bangkok, Thailand, 2003–2007.

Frits van Griensven; Anchalee Varangrat; Wipas Wimonsate; Suvimon Tanpradech; Keratikarn Kladsawad; Tareerat Chemnasiri; Orapin Suksripanich; Praphan Phanuphak; Philip Mock MappStats; Kamolset Kanggarnrua; Janet M. McNicholl; Tanarak Plipat

Background:Men who have sex with men (MSM) continue to be at high risk for HIV infection. Here we evaluate trends in HIV prevalence, estimated HIV incidence, and risk behavior among MSM in Bangkok, Thailand. Methods:Between 2003 and 2007, 3 biennial cross-sectional HIV prevalence assessments were conducted among MSM in Bangkok, Thailand, using venue-day-time sampling. Oral fluid was tested for HIV infection; demographic and behavioral data were self-collected using hand-held computers. Estimates of annual HIV incidence in young MSM were derived as follows: (number of HIV infections/sum of [current age-age at start of anal intercourse]) × 100). Logistic and Poisson regression was used to evaluate trends in HIV prevalence, estimated HIV incidence, and risk behavior. Findings:The overall HIV prevalence increased from 17.3% in 2003 to 28.3% in 2005 to 30.8% in 2007 (P < 0.001 for trend). The estimated HIV incidence among young MSM increased from 4.1% in 2003 to 6.4% in 2005, to 7.7% in 2007 (P < 0.02 for trend). The increase in HIV prevalence from 2005 to 2007 was not statistically significant. The proportion of men reporting anal sex and casual or steady male sex partners in the past 3 months significantly decreased, whereas the proportion reporting drug use and drug use during sex significantly increased. No increase was observed in the proportion of men reporting consistent condom use. Interpretation:Our data suggest that after a strong increase from 2003 to 2005, the HIV prevalence among MSM in Bangkok may have begun to stabilize. Given the continuing high levels of risk behavior and the estimated high HIV incidence in young MSM, additional HIV preventive interventions are necessary.


Journal of Acquired Immune Deficiency Syndromes | 2011

High susceptibility to repeated, low-dose, vaginal SHIV exposure late in the luteal phase of the menstrual cycle of pigtail macaques.

Vishwanathan Sa; Patricia C. Guenthner; Lin Cy; Charles Dobard; Sharma S; Debra R. Adams; Ron A. Otten; Walid Heneine; Hendry Rm; Janet M. McNicholl; Ellen N. Kersh

Fluctuations in susceptibility to HIV or SHIV during the menstrual cycle are currently not fully documented. To address this, the time point of infection was determined in 19 adult female pigtail macaques vaginally challenged during their undisturbed menstrual cycles with repeated, low-dose SHIVSF162P3 exposures. Eighteen macaques (95%) first displayed viremia in the follicular phase, as compared with 1 macaque (5%) in the luteal phase (P < 0.0001). Due to a viral eclipse phase, we estimated a window of most frequent virus transmission between days 24 and 31 of the menstrual cycle, in the late luteal phase. Thus, susceptibility to vaginal SHIV infection is significantly elevated in the second half of the menstrual cycle when progesterone levels are high and when local immunity may be low. Such susceptibility windows have been postulated before but not definitively documented. Our data support the findings of higher susceptibility to HIV in women during progesterone-dominated periods including pregnancy and contraceptive use.


Journal of Acquired Immune Deficiency Syndromes | 2008

Suppressive acyclovir therapy reduces HIV cervicovaginal shedding in HIV- and HSV-2-infected women, Chiang Rai, Thailand.

Eileen F. Dunne; Sara Whitehead; Maya Sternberg; Sukhon Thepamnuay; Wanna Leelawiwat; Janet M. McNicholl; Surin Sumanapun; Jordan W. Tappero; Taweesap Siriprapasiri; Lauri E. Markowitz

Background:Herpes simplex virus type 2 infection is important in the HIV epidemic and may contribute to increased HIV transmission. We evaluated the effect of suppressive acyclovir therapy on cervicovaginal HIV-1 shedding. Methods:HIV-1- and herpes simplex virus type 2-coinfected women aged 18-49 years with CD4 counts >200 cells/μL were enrolled in a randomized crossover trial of suppressive acyclovir therapy (NCT00362596, http://www.clinicaltrials.gov). For each woman, monthly plasma and weekly cervicovaginal lavage specimens were collected; the mean of the monthly median cervicovaginal lavage HIV-1 viral load and plasma HIV-1 viral load was compared. Results:Sixty-seven women were enrolled; at baseline, median CD4 count was 366 cells/μL, and median HIV-1 plasma viral load was 4.6 log10 copies/mL. The mean cervicovaginal lavage HIV-1 viral load was 1.9 (SD 0.8) log10 copies/mL during the acyclovir month and 2.2 (SD 0.7) log10 copies/mL during the placebo month (P < 0.0001); the mean decrease in HIV was 0.3 log10 copies/mL. The mean plasma HIV viral load during the acyclovir month (3.78 log10 copies/mL) was reduced compared with the placebo month (4.26 log10 copies/mL, P < 0.001). Conclusions:Acyclovir reduced HIV genital shedding and plasma viral load among HIV-1- and herpes simplex virus type 2-coinfected women. Further data from clinical trials will examine the effect of suppressive therapy on HIV transmission.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Intravaginal ring eluting tenofovir disoproxil fumarate completely protects macaques from multiple vaginal simian-HIV challenges

James M. Smith; Rachna Rastogi; Ryan S. Teller; Priya Srinivasan; Pedro M. M. Mesquita; Umadevi Nagaraja; Janet M. McNicholl; R. Michael Hendry; Chuong Dinh; Amy Martin; Betsy C. Herold; Patrick F. Kiser

Significance Topical prevention of HIV is designed to pharmacologically interrupt sexual transmission at the genital mucosa. Attempts at preventing transmission in women using vaginal gels have yielded disappointing results in part because of poor rates of adherence. Controlled topical drug delivery using intravaginal ring technology should improve efficacy and adherence by providing sustained mucosal delivery of antiretrovirals. In this paper, we describe a reservoir intravaginal ring that delivers tenofovir disoproxil fumarate (TDF) for 1 month. The ring protected pigtailed macaques from weekly vaginal simian–human immunodeficiency virus challenges for 4 mo. The sterilizing performance of this drug delivery system supports the concept that an intravaginal ring delivering TDF could be an effective tool for prevention of HIV sexual transmission in women. Topical preexposure prophylaxis interrupts HIV transmission at the site of mucosal exposure. Intermittently dosed vaginal gels containing the HIV-1 reverse transcriptase inhibitor tenofovir protected pigtailed macaques depending on the timing of viral challenge relative to gel application. However, modest or no protection was observed in clinical trials. Intravaginal rings (IVRs) may improve efficacy by providing long-term sustained drug delivery leading to constant mucosal antiretroviral concentrations and enhancing adherence. Although a few IVRs have entered the clinical pipeline, 100% efficacy in a repeated macaque vaginal challenge model has not been achieved. Here we describe a reservoir IVR technology that delivers the tenofovir prodrug tenofovir disoproxil fumarate (TDF) continuously over 28 d. With four monthly ring changes in this repeated challenge model, TDF IVRs generated reproducible and protective drug levels. All TDF IVR-treated macaques (n = 6) remained seronegative and simian-HIV RNA negative after 16 weekly vaginal exposures to 50 tissue culture infectious dose SHIV162p3. In contrast, 11/12 control macaques became infected, with a median of four exposures assuming an eclipse of 7 d from infection to virus RNA detection. Protection was associated with tenofovir levels in vaginal fluid [mean 1.8 × 105 ng/mL (range 1.1 × 104 to 6.6 × 105 ng/mL)] and ex vivo antiviral activity of cervicovaginal lavage samples. These observations support further advancement of TDF IVRs as well as the concept that extended duration drug delivery devices delivering topical antiretrovirals could be effective tools in preventing the sexual transmission of HIV in humans.


Nature Medicine | 2014

Targeting α4β7 integrin reduces mucosal transmission of simian immunodeficiency virus and protects gut-associated lymphoid tissue from infection.

Siddappa N. Byrareddy; Brianne Kallam; James Arthos; Claudia Cicala; Fatima Nawaz; Joseph Hiatt; Ellen N. Kersh; Janet M. McNicholl; Debra L. Hanson; Keith A. Reimann; Markus Brameier; Lutz Walter; Kenneth Rogers; Ann E. Mayne; Paul Dunbar; Tara Villinger; Dawn M. Little; Tristram G. Parslow; Philip J. Santangelo; Francois Villinger; Anthony S. Fauci; Aftab A. Ansari

α4β7 integrin expressing CD4+ T cells preferentially traffic to gut-associated lymphoid tissues (GALT) and play a key role in HIV/SIV pathogenesis. The administration of an anti-α4β7 monoclonal antibody during acute infection protects macaques from transmission following repeated low-dose intra-vaginal challenges with SIVmac251. In treated animals that became infected the GALT was significantly protected and CD4+ T–cell numbers were maintained. Thus, targeting α4β7 reduces mucosal transmission of SIV in macaques.α4β7 integrin–expressing CD4+ T cells preferentially traffic to gut-associated lymphoid tissue (GALT) and have a key role in HIV and simian immunodeficiency virus (SIV) pathogenesis. We show here that the administration of an anti-α4β7 monoclonal antibody just prior to and during acute infection protects rhesus macaques from transmission following repeated low-dose intravaginal challenges with SIVmac251. In treated animals that became infected, the GALT was significantly protected from infection and CD4+ T cell numbers were maintained in both the blood and the GALT. Thus, targeting α4β7 reduces mucosal transmission of SIV in macaques.


AIDS Research and Human Retroviruses | 2001

An HLA-Directed Molecular and Bioinformatics Approach Identifies New HLA-A11 HIV-1 Subtype E Cytotoxic T Lymphocyte Epitopes in HIV-1-Infected Thais

Kyle Bond; Busarawan Sriwanthana; Thomas W. Hodge; Anne S. De Groot; Timothy D. Mastro; Nancy L. Young; Nattawan Promadej; John D. Altman; Khanchit Limpakarnjanarat; Janet M. McNicholl

Only limited cytotoxic T lymphocyte (CTL) epitope mapping has been done in nonsubtype B HIV-infected persons. We used molecular immunogenetic tools to determine HIV-specific CTL responses in HIV-1 Env subtype E-infected female sex workers (FSWs) from northern Thailand, where more than 50% of the population is HLA-A11 positive. EpiMatrix, a computer-based T cell epitope prediction algorithm, and a manual editing approach were used to predict 77 possible HLA-A11 CTL epitopes in HIV-1, some of which were conserved between subtypes B and E. MHC binding of these peptides was determined in an HLA-A11 stabilization assay, and binding peptides were tested for CTL recognition in eight HLA-A11-positive FSWs. Subtype E versions of known HLA-A2 subtype B HIV epitopes were also tested in four HLA-A2 positive FSWs. CTL responses were detected in all HLA-A11-positive and in three of four HLA-A2-positive persons. Among the 12 FSWs responses to peptides were found to Pol in 9 (75%), Env in 7 (58%), Nef in 5 (42%), and Gag in 5 (42%), and to conserved epitopes in 8 (67%). To identify HLA-A11 CTL epitopes in the absence of prediction tools, it would have been necessary to test almost 3000 10-mer peptides. EpiMatrix and manual predictions reduced this number to 77, of which 26 were MHC binding and 12 were CTL epitopes. Six of these HLA-A11 CTL epitopes have not been previously reported and are located in RT, gp120, and gp41. This report of CTL responses in subtype E-infected individuals defines epitopes that may be useful in HIV pathogenesis or vaccine studies.

Collaboration


Dive into the Janet M. McNicholl's collaboration.

Top Co-Authors

Avatar

Ellen N. Kersh

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

James M. Smith

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Kachit Choopanya

Bangkok Metropolitan Administration

View shared research outputs
Top Co-Authors

Avatar

Katherine Butler

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Suphak Vanichseni

Bangkok Metropolitan Administration

View shared research outputs
Top Co-Authors

Avatar

Jordan W. Tappero

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Marcel E. Curlin

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

R. Michael Hendry

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Sundaram A. Vishwanathan

Centers for Disease Control and Prevention

View shared research outputs
Researchain Logo
Decentralizing Knowledge